Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
Add more filters










Publication year range
1.
Adv Sci (Weinh) ; : e2400437, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38885417

ABSTRACT

SH2B1 mutations are associated with obesity, type 2 diabetes, and metabolic dysfunction-associated steatotic liver disease (MASLD) in humans. Global deletion of Sh2b1 results in severe obesity, type 2 diabetes, and MASLD in mice. Neuron-specific restoration of SH2B1 rescues the obesity phenotype of Sh2b1-null mice, indicating that the brain is a main SH2B1 target. However, SH2B1 neurocircuits remain elusive. SH2B1-expressing neurons in the paraventricular hypothalamus (PVHSH2B1) and a PVHSH2B1→dorsal raphe nucleus (DRN) neurocircuit are identified here. PVHSH2B1 axons monosynaptically innervate DRN neurons. Optogenetic stimulation of PVHSH2B1 axonal fibers in the DRN suppresses food intake. Chronic inhibition of PVHSH2B1 neurons causes obesity. In male and female mice, either embryonic-onset or adult-onset deletion of Sh2b1 in PVH neurons causes energy imbalance, obesity, insulin resistance, glucose intolerance, and MASLD. Ablation of Sh2b1 in the DRN-projecting PVHSH2B1 subpopulation also causes energy imbalance, obesity, and metabolic disorders. Conversely, SH2B1 overexpression in either total or DRN-projecting PVHSH2B1 neurons protects against diet-induced obesity. SH2B1 binds to TrkB and enhances brain-derived neurotrophic factor (BDNF) signaling. Ablation of Sh2b1 in PVHSH2B1 neurons induces BDNF resistance in the PVH, contributing to obesity. In conclusion, these results unveil a previously unrecognized PVHSH2B1→DRN neurocircuit through which SH2B1 defends against obesity by enhancing BDNF/TrkB signaling.

2.
Nat Cell Biol ; 26(6): 917-931, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38714852

ABSTRACT

Upon endoplasmic reticulum (ER) stress, activation of the ER-resident transmembrane protein kinase/endoribonuclease inositol-requiring enzyme 1 (IRE1) initiates a key branch of the unfolded protein response (UPR) through unconventional splicing generation of the transcription factor X-box-binding protein 1 (XBP1s). Activated IRE1 can form large clusters/foci, whose exact dynamic architectures and functional properties remain largely elusive. Here we report that, in mammalian cells, formation of IRE1α clusters is an ER membrane-bound phase separation event that is coupled to the assembly of stress granules (SGs). In response to different stressors, IRE1α clusters are dynamically tethered to SGs at the ER. The cytosolic linker portion of IRE1α possesses intrinsically disordered regions and is essential for its condensation with SGs. Furthermore, disruption of SG assembly abolishes IRE1α clustering and compromises XBP1 mRNA splicing, and such IRE1α-SG coalescence engenders enrichment of the biochemical components of the pro-survival IRE1α-XBP1 pathway during ER stress. Our findings unravel a phase transition mechanism for the spatiotemporal assembly of IRE1α-SG condensates to establish a more efficient IRE1α machinery, thus enabling higher stress-handling capacity.


Subject(s)
Endoplasmic Reticulum Stress , Endoribonucleases , Protein Serine-Threonine Kinases , X-Box Binding Protein 1 , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Endoribonucleases/metabolism , Endoribonucleases/genetics , Humans , X-Box Binding Protein 1/metabolism , X-Box Binding Protein 1/genetics , Animals , RNA Splicing , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/genetics , Stress Granules/metabolism , Stress Granules/genetics , Regulatory Factor X Transcription Factors/metabolism , Regulatory Factor X Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Unfolded Protein Response , Mice , HeLa Cells , Cytoplasmic Granules/metabolism , Cytoplasmic Granules/genetics , Signal Transduction
3.
Cell Rep ; 43(3): 113875, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38451818

ABSTRACT

Liver injury stimulates hepatocyte replication and hepatic stellate cell (HSC) activation, thereby driving liver regeneration. Aberrant HSC activation induces liver fibrosis. However, mechanisms underlying liver regeneration and fibrosis remain poorly understood. Here, we identify hepatic Snai1 and Snai2 as important transcriptional regulators for liver regeneration and fibrosis. Partial hepatectomy or CCl4 treatment increases occupancies of Snai1 and Snai2 on cyclin A2 and D1 promoters in the liver. Snai1 and Snai2 in turn increase promoter H3K27 acetylation and cyclin A2/D1 expressions. Hepatocyte-specific deletion of both Snai1 and Snai2, but not one alone, suppresses liver cyclin A2/D1 expression and regenerative hepatocyte proliferation after hepatectomy or CCl4 treatments but augments CCl4-stimulated HSC activation and liver fibrosis. Conversely, Snai2 overexpression in the liver enhances hepatocyte replication and suppresses liver fibrosis after CCl4 treatment. These results suggest that hepatic Snai1 and Snai2 directly promote, via histone modifications, reparative hepatocyte replication and indirectly inhibit liver fibrosis.


Subject(s)
Cyclin A2 , Liver Regeneration , Animals , Mice , Cyclin A2/metabolism , Hepatectomy , Liver/metabolism , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Liver Regeneration/physiology
5.
Adv Sci (Weinh) ; 10(28): e2301645, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37526326

ABSTRACT

White adipose tissue (WAT) lipolysis releases free fatty acids as a key energy substance to support metabolism in fasting, cold exposure, and exercise. Atgl, in concert with Cgi-58, catalyzes the first lipolytic reaction. The sympathetic nervous system (SNS) stimulates lipolysis via neurotransmitter norepinephrine that activates adipocyte ß adrenergic receptors (Adrb1-3). In obesity, adipose Adrb signaling and lipolysis are impaired, contributing to pathogenic WAT expansion; however, the underling mechanism remains poorly understood. Recent studies highlight importance of N6 -methyladenosine (m6A)-based RNA modification in health and disease. METTL14 heterodimerizes with METTL3 to form an RNA methyltransferase complex that installs m6A in transcripts. Here, this work shows that adipose Mettl3 and Mettl14 are influenced by fasting, refeeding, and insulin, and are upregulated in high fat diet (HFD) induced obesity. Adipose Adrb2, Adrb3, Atgl, and Cgi-58 transcript m6A contents are elevated in obesity. Mettl14 ablation decreases these transcripts' m6A contents and increases their translations and protein levels in adipocytes, thereby increasing Adrb signaling and lipolysis. Mice with adipocyte-specific deletion of Mettl14 are resistant to HFD-induced obesity, insulin resistance, glucose intolerance, and nonalcoholic fatty liver disease (NAFLD). These results unravel a METTL14/m6A/translation pathway governing Adrb signaling and lipolysis. METTL14/m6A-based epitranscriptomic reprogramming impairs adipose Adrb signaling and lipolysis, promoting obesity, NAFLD, and metabolic disease.


Subject(s)
Insulin Resistance , Non-alcoholic Fatty Liver Disease , Animals , Mice , Adrenergic Agents , Lipolysis/physiology , Methyltransferases/genetics , Methyltransferases/metabolism , Obesity/metabolism , RNA/metabolism
6.
J Clin Invest ; 133(4)2023 02 15.
Article in English | MEDLINE | ID: mdl-36512408

ABSTRACT

Leptin exerts its biological actions by activating the long-form leptin receptor (LepRb). LepRb signaling impairment and leptin resistance are believed to cause obesity. The transcription factor Slug - also known as Snai2 - recruits epigenetic modifiers and regulates gene expression by an epigenetic mechanism; however, its epigenetic action has not been explored in leptin resistance. Here, we uncover a proobesity function of neuronal Slug. Hypothalamic Slug was upregulated in obese mice. LepRb+ cell-specific Slug-knockout (SlugΔLepRb) mice were resistant to diet-induced obesity, type 2 diabetes, and liver steatosis and experienced decreased food intake and increased fat thermogenesis. Leptin stimulated hypothalamic Stat3 phosphorylation and weight loss to a markedly higher level in SlugΔLepRb than in Slugfl/fl mice, even before their body weight divergence. Conversely, hypothalamic LepRb+ neuron-specific overexpression of Slug, mediated by AAV-hSyn-DIO-Slug transduction, induced leptin resistance, obesity, and metabolic disorders in mice on a chow diet. At the genomic level, Slug bound to and repressed the LepRb promoter, thereby inhibiting LepRb transcription. Consistently, Slug deficiency decreased methylation of LepRb promoter H3K27, a repressive epigenetic mark, and increased LepRb mRNA levels in the hypothalamus. Collectively, these results unravel what we believe to be a previously unrecognized hypothalamic neuronal Slug/epigenetic reprogramming/leptin resistance axis that promotes energy imbalance, obesity, and metabolic disease.


Subject(s)
Non-alcoholic Fatty Liver Disease , Obesity , Receptors, Leptin , Snail Family Transcription Factors , Animals , Mice , Diabetes Mellitus, Type 2/metabolism , Hypothalamus/metabolism , Leptin/genetics , Leptin/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Obesity/genetics , Obesity/metabolism , Receptors, Leptin/genetics , Receptors, Leptin/metabolism , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism
7.
Nat Metab ; 4(9): 1166-1184, 2022 09.
Article in English | MEDLINE | ID: mdl-36123394

ABSTRACT

Adipose tissue undergoes thermogenic remodeling in response to thermal stress and metabolic cues, playing a crucial role in regulating energy expenditure and metabolic homeostasis. Endoplasmic reticulum (ER) stress is associated with adipose dysfunction in obesity and metabolic disease. It remains unclear, however, if ER stress-signaling in adipocytes mechanistically mediates dysregulation of thermogenic fat. Here we show that inositol-requiring enzyme 1α (IRE1α), a key ER stress sensor and signal transducer, acts in both white and beige adipocytes to impede beige fat activation. Ablation of adipocyte IRE1α promotes browning/beiging of subcutaneous white adipose tissue following cold exposure or ß3-adrenergic stimulation. Loss of IRE1α alleviates diet-induced obesity and augments the anti-obesity effect of pharmacologic ß3-adrenergic stimulation. Notably, IRE1α suppresses stimulated lipolysis and degrades Ppargc1a messenger RNA through its RNase activity to downregulate the thermogenic gene program. Hence, blocking IRE1α bears therapeutic potential in unlocking adipocytes' thermogenic capacity to combat obesity and metabolic disorders.


Subject(s)
Endoribonucleases , Inositol , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Protein Serine-Threonine Kinases , Adipocytes/metabolism , Adrenergic Agents/pharmacology , Animals , Endoribonucleases/genetics , Endoribonucleases/metabolism , Inositol/pharmacology , Mice , Obesity/genetics , Obesity/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA Stability , RNA, Messenger , Thermogenesis/genetics
8.
Nat Commun ; 13(1): 5111, 2022 08 30.
Article in English | MEDLINE | ID: mdl-36042192

ABSTRACT

Excessive cholangiocyte expansion (ductular reaction) promotes liver disease progression, but the underlying mechanism is poorly understood. Here we identify biliary NF-κB-inducing kinase (NIK) as a pivotal regulator of ductular reaction. NIK is known to activate the noncanonical IKKα/NF-κB2 pathway and regulate lymphoid tissue development. We find that cholangiocyte NIK is upregulated in mice with cholestasis induced by bile duct ligation (BDL), 5-diethoxycarbonyl-1,4-dihydrocollidine (DDC), or α-naphtyl-isothiocyanate (ANIT). DDC, ANIT, or BDL induces ductular reaction, liver injury, inflammation, and fibrosis in mice. Cholangiocyte-specific deletion of NIK, but not IKKα, blunts these pathological alterations. NIK inhibitor treatment similarly ameliorates DDC-induced ductular reaction, liver injury, and fibrosis. Biliary NIK directly increases cholangiocyte proliferation while suppressing cholangiocyte death, and it also promotes secretion of cholangiokines from cholangiocytes. Cholangiokines stimulate liver macrophages and hepatic stellate cells, augmenting liver inflammation and fibrosis. These results unveil a NIK/ductular reaction axis and a NIK/cholangiokine axis that promote liver disease progression.


Subject(s)
Liver Diseases , Liver , Protein Serine-Threonine Kinases/metabolism , Animals , Disease Progression , Fibrosis , Liver/metabolism , Liver Cirrhosis/pathology , Liver Diseases/pathology , Mice , NF-kappaB-Inducing Kinase
9.
Annu Rev Nutr ; 42: 91-113, 2022 08 22.
Article in English | MEDLINE | ID: mdl-35584814

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD), a spectrum of metabolic liver disease associated with obesity, ranges from relatively benign hepatic steatosis to nonalcoholic steatohepatitis (NASH). The latter is characterized by persistent liver injury, inflammation, and liver fibrosis, which collectively increase the risk for end-stage liver diseases such as cirrhosis and hepatocellular carcinoma. Recent work has shed new light on the pathophysiology of NAFLD/NASH, particularly the role of genetic, epigenetic, and dietary factors and metabolic dysfunctions in other tissues in driving excess hepatic fat accumulation and liver injury. In parallel, single-cell RNA sequencing studies have revealed unprecedented details of the molecular nature of liver cell heterogeneity, intrahepatic cross talk, and disease-associated reprogramming of the liver immune and stromal vascular microenvironment. This review covers the recent advances in these areas, the emerging concepts of NASH pathogenesis, and potential new therapeutic opportunities.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Non-alcoholic Fatty Liver Disease , Carcinoma, Hepatocellular/complications , Carcinoma, Hepatocellular/pathology , Humans , Liver/metabolism , Liver Cirrhosis/complications , Liver Cirrhosis/pathology , Liver Neoplasms/complications , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Non-alcoholic Fatty Liver Disease/metabolism , Tumor Microenvironment
10.
Redox Biol ; 52: 102308, 2022 06.
Article in English | MEDLINE | ID: mdl-35390677

ABSTRACT

The incidence of Parkinson's disease (PD) has increased tremendously, especially in the aged population and people with metabolic dysfunction; however, its underlying molecular mechanisms remain unclear. SH2B1, an intracellular adaptor protein, contributes to the signal transduction of several receptor tyrosine kinases and exerts beneficial metabolic effects for body weight regulation; however, whether SH2B1 plays a major role in pathological neurodegeneration in PD has not yet been investigated. This study aimed to investigate the effects of SH2B1 in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mice with Sh2b1 deficiency or neuron-specific Sh2b1 overexpression. Cellular and molecular mechanisms were elucidated using human dopaminergic neuron SH-SY5Y cells analysed. We found that SH2B1 expression was confirmed to be downregulated in the blood samples of PD patients and in the brains of mice with MPTP-induced chronic PD. Sh2b1 deficiency caused marked exacerbation of behavioural defects and increased neuronal apoptosis in MPTP-treated mice, whereas restoration of neuron-specific Sh2b1 expression significantly reversed these effects. Similar results were observed in MPP + -treated SH-SY5Y cells. Mechanistically, upon binding to heat shock cognate 70 (HSC70), SH2B1 promotes HSC70-related recognition and PLIN4 lysosomal translocation and degradation, thus suppressing lipid peroxidation stress in the brains of PD mice. Adeno-associated virus-mediated rescue of neuronal HSC70 expression functionally alleviated the neuropathology of PD in wild-type but not in Sh2b1-deficient mice. This is the first study to examine the molecular underpinnings of SH2B1 against MPTP-induced neurodegeneration through cell autonomous promotion of neuronal survival in an in vivo PD model. Our findings reveal that SH2B1 antagonizes neurodegenerative pathology in PD via the SH2B1-HSC70-PLIN4 axis.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Adaptor Proteins, Signal Transducing/metabolism , Parkinson Disease , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/adverse effects , Adaptor Proteins, Signal Transducing/genetics , Aged , Animals , Apoptosis , Disease Models, Animal , Dopaminergic Neurons/metabolism , Humans , Mice , Mice, Inbred C57BL , Parkinson Disease/genetics , Parkinson Disease/metabolism , Perilipin-4/metabolism
11.
Blood ; 139(16): 2547-2552, 2022 04 21.
Article in English | MEDLINE | ID: mdl-34990508

ABSTRACT

Intestinal iron absorption is activated during increased systemic demand for iron. The best-studied example is iron deficiency anemia, which increases intestinal iron absorption. Interestingly, the intestinal response to anemia is very similar to that of iron overload disorders, as both the conditions activate a transcriptional program that leads to a hyperabsorption of iron via the transcription factor hypoxia-inducible factor 2α (HIF2α). However, pathways for selective targeting of intestine-mediated iron overload remain unknown. Nuclear receptor coactivator 4 (NCOA4) is a critical cargo receptor for autophagic breakdown of ferritin and the subsequent release of iron, in a process termed ferritinophagy. Our work demonstrates that NCOA4-mediated intestinal ferritinophagy is integrated into systemic iron demand via HIF2α. To demonstrate the importance of the intestinal HIF2α/ferritinophagy axis in systemic iron homeostasis, whole-body and intestine-specific NCOA4-/- mouse lines were generated and assessed. The analyses revealed that the intestinal and systemic response to iron deficiency was not altered after disruption of intestinal NCOA4. However, in a mouse model of hemochromatosis, ablation of intestinal NCOA4 was protective against iron overload. Therefore, NCOA4 can be selectively targeted for the management of iron overload disorders without disrupting the physiological processes involved in the response to systemic iron deficiency.


Subject(s)
Anemia , Basic Helix-Loop-Helix Transcription Factors/metabolism , Hemochromatosis , Iron Overload , Animals , Enterocytes/metabolism , Hemochromatosis/genetics , Iron/metabolism , Mice , Nuclear Receptor Coactivators/genetics , Transcription Factors/metabolism
12.
Hepatol Commun ; 5(10): 1704-1720, 2021 10.
Article in English | MEDLINE | ID: mdl-34558831

ABSTRACT

Drug-induced hepatotoxicity limits development of new effective medications. Drugs and numerous endogenous/exogenous agents are metabolized/detoxified by hepatocytes, during which reactive oxygen species (ROS) are generated as a by-product. ROS has broad adverse effects on liver function and integrity, including damaging hepatocyte proteins, lipids, and DNA and promoting liver inflammation and fibrosis. ROS in concert with iron overload drives ferroptosis. Hepatic nuclear factor kappa B (NF-κB)-inducing kinase (NIK) is aberrantly activated in a broad spectrum of liver disease. NIK phosphorylates and activates inhibitor of NF-κB kinase subunit alpha (IKKα), and the hepatic NIK/IKKα cascade suppresses liver regeneration. However, the NIK/IKKα pathway has not been explored in drug-induced liver injury. Here, we identify hepatic NIK as a previously unrecognized mediator for acetaminophen (APAP)-induced acute liver failure. APAP treatment increased both NIK transcription and NIK protein stability in primary hepatocytes as well as in liver in mice. Hepatocyte-specific overexpression of NIK augmented APAP-induced liver oxidative stress in mice and increased hepatocyte death and mortality in a ROS-dependent manner. Conversely, hepatocyte-specific ablation of NIK or IKKα mitigated APAP-elicited hepatotoxicity and mortality. NIK increased lipid peroxidation and cell death in APAP-stimulated primary hepatocytes. Pretreatment with antioxidants or ferroptosis inhibitors blocked NIK/APAP-induced hepatocyte death. Conclusion: We unravel a previously unrecognized NIK/IKKα/ROS/ferroptosis axis engaged in liver disease progression.


Subject(s)
Chemical and Drug Induced Liver Injury/etiology , Ferroptosis/drug effects , Oxidative Stress/drug effects , Protein Kinase Inhibitors/metabolism , Protein Serine-Threonine Kinases/metabolism , Acetaminophen/adverse effects , Animals , Hepatocytes/drug effects , Lipid Peroxidation/drug effects , Liver/drug effects , Liver Regeneration/drug effects , Mice , Phosphorylation/drug effects , Reactive Oxygen Species/metabolism , NF-kappaB-Inducing Kinase
13.
Nat Commun ; 12(1): 2621, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33976147

ABSTRACT

Cullin-RING E3 ligases (CRLs) regulate the turnover of approximately 20% of mammalian cellular proteins. Neddylation of individual cullin proteins is essential for the activation of each CRL. We report herein the discovery of DI-1548 and DI-1859 as two potent, selective and covalent DCN1 inhibitors. These inhibitors selectively inhibit neddylation of cullin 3 in cells at low nanomolar concentrations and are 2-3 orders of magnitude more potent than our previously reported reversible DCN1 inhibitor. Mass spectrometric analysis and co-crystal structures reveal that these compounds employ a unique mechanism of covalent bond formation with DCN1. DI-1859 induces a robust increase of NRF2 protein, a CRL3 substrate, in mouse liver and effectively protects mice from acetaminophen-induced liver damage. Taken together, this study demonstrates the therapeutic potential of selective inhibition of cullin neddylation.


Subject(s)
Acetaminophen/toxicity , Chemical and Drug Induced Liver Injury/prevention & control , Cullin Proteins/metabolism , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Protective Agents/pharmacology , Acetaminophen/administration & dosage , Animals , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/pathology , Crystallography, X-Ray , Disease Models, Animal , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Liver/drug effects , Liver/pathology , Male , Mice , NEDD8 Protein/metabolism , NF-E2-Related Factor 2/metabolism , Protective Agents/chemistry , Protective Agents/therapeutic use , Protein Processing, Post-Translational/drug effects
14.
Am J Respir Cell Mol Biol ; 65(2): 214-221, 2021 08.
Article in English | MEDLINE | ID: mdl-33891828

ABSTRACT

Obesity elevates the plasma level of leptin, which has been associated with hypertension. Our recent studies in mice demonstrated that leptin increases blood pressure by activating the carotid sinus nerve, which transmits the chemosensory input from carotid bodies (CBs) to the medullary centers, and that the effect of leptin is mediated via Trpm7 (TRP [transient receptor potential] melastatin 7) channels in CB glomus cells. We also found that Trpm7 overexpression and Trpm7 promoter demethylation in CBs correlate positively with the hyperleptinemia and leptin receptor overexpression in CBs. Hence, we postulated that leptin epigenetically regulates Trpm7 expression in CBs. We addressed our hypothesis by using rat adrenal pheochromocytoma (PC12) cells as a model of CB glomus cells. PC12 cells expressing LEPRb (long, active form of leptin receptor) showed dramatic induction of the promoter activity and expression of Trpm7 upon leptin treatment. The increased Trpm7 expression coincided with the reduction of CpG site-specific methylation and trimethylation of H3K27 (H3 [histone 3] K27 [lysine 27]) and the increase of acetylation of H3K27 and trimethylation of H3K4 (H3 lysine 4) at the Trpm7 promoter. The inhibitor of STAT3 (signal transducer and activator of transcription 3) signaling, SD1008, reversed the leptin-induced Trpm7 promoter activity via modulations of the binding of pSTAT3 (phosphorylated STAT3) and DNMT3B (DNA methyltransferase 3B) and modifications of H3K27 and H3K4 at the Trpm7 promoter. Our results suggest that leptin-activated pSTAT3 epigenetically regulates the transcription of Trpm7 through DNA methylation and histone modifications. Because epigenetic changes are reversible, targeting epigenetic modifications of Trpm7 may serve as a new therapeutic approach for the treatment of hypertension in obesity.


Subject(s)
Adrenal Gland Neoplasms/metabolism , Epigenesis, Genetic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Leptin/pharmacology , Neoplasm Proteins/biosynthesis , Pheochromocytoma/metabolism , TRPM Cation Channels/biosynthesis , Adrenal Gland Neoplasms/genetics , Adrenal Gland Neoplasms/pathology , Animals , Neoplasm Proteins/genetics , PC12 Cells , Pheochromocytoma/genetics , Pheochromocytoma/pathology , Rats , TRPM Cation Channels/genetics
15.
J Biol Chem ; 296: 100235, 2021.
Article in English | MEDLINE | ID: mdl-33376138

ABSTRACT

Epigenetic mechanisms that alter heritable gene expression and chromatin structure play an essential role in many biological processes, including liver function. Human MOF (males absent on the first) is a histone acetyltransferase that is globally downregulated in human steatohepatitis. However, the function of MOF in the liver remains unclear. Here, we report that MOF plays an essential role in adult liver. Genetic deletion of Mof by Mx1-Cre in the liver leads to acute liver injury, with increase of lipid deposition and fibrosis akin to human steatohepatitis. Surprisingly, hepatocyte-specific Mof deletion had no overt liver abnormality. Using the in vitro coculturing experiment, we show that Mof deletion-induced liver injury requires coordinated changes and reciprocal signaling between hepatocytes and Kupffer cells, which enables feedforward regulation to augment inflammation and apoptotic responses. At the molecular level, Mof deletion induced characteristic changes in metabolic gene programs, which bore noticeable similarity to the molecular signature of human steatohepatitis. Simultaneous deletion of Mof in both hepatocytes and macrophages results in enhanced expression of inflammatory genes and NO signaling in vitro. These changes, in turn, lead to apoptosis of hepatocytes and lipotoxicity. Our work highlights the importance of histone acetyltransferase MOF in maintaining metabolic liver homeostasis and sheds light on the epigenetic dysregulation in liver pathogenesis.


Subject(s)
Histone Acetyltransferases/genetics , Inflammation/metabolism , Liver Diseases/genetics , Liver/injuries , Nitric Oxide/genetics , Apoptosis/genetics , Chromatin/genetics , Epigenesis, Genetic/genetics , Fatty Liver/genetics , Fatty Liver/metabolism , Fatty Liver/pathology , Gene Deletion , Gene Expression Regulation/genetics , Hepatocytes/metabolism , Hepatocytes/pathology , Histone Acetyltransferases/chemistry , Humans , Inflammation/genetics , Inflammation/pathology , Lipids/adverse effects , Lipids/genetics , Liver/metabolism , Liver/pathology , Liver Diseases/metabolism , Liver Diseases/pathology , Macrophages/metabolism , Macrophages/pathology , Nitric Oxide/metabolism , Signal Transduction/genetics
16.
Nat Commun ; 11(1): 5310, 2020 Oct 15.
Article in English | MEDLINE | ID: mdl-33060593

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

17.
FASEB J ; 34(11): 14863-14877, 2020 11.
Article in English | MEDLINE | ID: mdl-32918517

ABSTRACT

Appropriate control of hepatic gluconeogenesis is essential for the organismal survival upon prolonged fasting and maintaining systemic homeostasis under metabolic stress. Here, we show protein arginine methyltransferase 1 (PRMT1), a key enzyme that catalyzes the protein arginine methylation process, particularly the isoform encoded by Prmt1 variant 2 (PRMT1V2), is critical in regulating gluconeogenesis in the liver. Liver-specific deletion of Prmt1 reduced gluconeogenic capacity in cultured hepatocytes and in the liver. Prmt1v2 was expressed at a higher level compared to Prmt1v1 in hepatic tissue and cells. Gain-of-function of PRMT1V2 clearly activated the gluconeogenic program in hepatocytes via interactions with PGC1α, a key transcriptional coactivator regulating gluconeogenesis, enhancing its activity via arginine methylation, while no effects of PRMT1V1 were observed. Similar stimulatory effects of PRMT1V2 in controlling gluconeogenesis were observed in human HepG2 cells. PRMT1, specifically PRMT1V2, was stabilized in fasted liver and hepatocytes treated with glucagon, in a PGC1α-dependent manner. PRMT1, particularly Prmt1v2, was significantly induced in the liver of streptozocin-induced type 1 diabetes and high fat diet-induced type 2 diabetes mouse models and liver-specific Prmt1 deficiency drastically ameliorated diabetic hyperglycemia. These findings reveal that PRMT1 modulates gluconeogenesis and mediates glucose homeostasis under physiological and pathological conditions, suggesting that deeper understanding how PRMT1 contributes to the coordinated efforts in glycemic control may ultimately present novel therapeutic strategies that counteracts hyperglycemia in disease settings.


Subject(s)
Gluconeogenesis , Hepatocytes/metabolism , Hyperglycemia/genetics , Protein-Arginine N-Methyltransferases/metabolism , Animals , Cells, Cultured , Gain of Function Mutation , Glucagon/metabolism , Glucose/metabolism , Hep G2 Cells , Humans , Hyperglycemia/metabolism , Mice , Mice, Inbred C57BL , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein-Arginine N-Methyltransferases/genetics
18.
J Clin Invest ; 130(6): 2992-3004, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32365055

ABSTRACT

De novo lipogenesis is tightly regulated by insulin and nutritional signals to maintain metabolic homeostasis. Excessive lipogenesis induces lipotoxicity, leading to nonalcoholic fatty liver disease (NAFLD) and type 2 diabetes. Genetic lipogenic programs have been extensively investigated, but epigenetic regulation of lipogenesis is poorly understood. Here, we identified Slug as an important epigenetic regulator of lipogenesis. Hepatic Slug levels were markedly upregulated in mice by either feeding or insulin treatment. In primary hepatocytes, insulin stimulation increased Slug expression, stability, and interactions with epigenetic enzyme lysine-specific demethylase-1 (Lsd1). Slug bound to the fatty acid synthase (Fasn) promoter where Slug-associated Lsd1 catalyzed H3K9 demethylation, thereby stimulating Fasn expression and lipogenesis. Ablation of Slug blunted insulin-stimulated lipogenesis. Conversely, overexpression of Slug, but not a Lsd1 binding-defective Slug mutant, stimulated Fasn expression and lipogenesis. Lsd1 inhibitor treatment also blocked Slug-stimulated lipogenesis. Remarkably, hepatocyte-specific deletion of Slug inhibited the hepatic lipogenic program and protected against obesity-associated NAFLD, insulin resistance, and glucose intolerance in mice. Conversely, liver-restricted overexpression of Slug, but not the Lsd1 binding-defective Slug mutant, had the opposite effects. These results unveil an insulin/Slug/Lsd1/H3K9 demethylation lipogenic pathway that promotes NAFLD and type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Epigenesis, Genetic , Lipogenesis , Non-alcoholic Fatty Liver Disease/metabolism , Snail Family Transcription Factors/biosynthesis , Animals , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Fatty Acid Synthase, Type I/genetics , Fatty Acid Synthase, Type I/metabolism , Gene Deletion , Hepatocytes , Histone Demethylases/genetics , Histone Demethylases/metabolism , Humans , Mice , Mice, Transgenic , Mutation , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , Snail Family Transcription Factors/genetics
19.
Nat Commun ; 11(1): 1517, 2020 03 23.
Article in English | MEDLINE | ID: mdl-32251290

ABSTRACT

Leptin stimulates the sympathetic nervous system (SNS), energy expenditure, and weight loss; however, the underlying molecular mechanism remains elusive. Here, we uncover Sh2b1 in leptin receptor (LepR) neurons as a critical component of a SNS/brown adipose tissue (BAT)/thermogenesis axis. LepR neuron-specific deletion of Sh2b1 abrogates leptin-stimulated sympathetic nerve activation and impairs BAT thermogenic programs, leading to reduced core body temperature and cold intolerance. The adipose SNS degenerates progressively in mutant mice after 8 weeks of age. Adult-onset ablation of Sh2b1 in the mediobasal hypothalamus also impairs the SNS/BAT/thermogenesis axis; conversely, hypothalamic overexpression of human SH2B1 has the opposite effects. Mice with either LepR neuron-specific or adult-onset, hypothalamus-specific ablation of Sh2b1 develop obesity, insulin resistance, and liver steatosis. In contrast, hypothalamic overexpression of SH2B1 protects against high fat diet-induced obesity and metabolic syndromes. Our results unravel an unrecognized LepR neuron Sh2b1/SNS/BAT/thermogenesis axis that combats obesity and metabolic disease.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Fatty Liver/pathology , Insulin Resistance , Neurons/metabolism , Obesity/pathology , Adaptor Proteins, Signal Transducing/genetics , Adipose Tissue, Brown/innervation , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/pathology , Animals , Diet, High-Fat/adverse effects , Disease Models, Animal , Fatty Liver/etiology , Female , Gene Knock-In Techniques , Gene Knockout Techniques , Humans , Hypothalamus/pathology , Leptin/metabolism , Liver/pathology , Male , Mice , Mice, Transgenic , Obesity/etiology , Receptors, Leptin/metabolism , Sympathetic Nervous System/physiology , Thermogenesis/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...